Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65.097
Filter
1.
Biomaterials ; 312: 122731, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39153324

ABSTRACT

Tumor-associated inflammation drives cancer progression and therapy resistance, often linked to the infiltration of monocyte-derived tumor-associated macrophages (TAMs), which are associated with poor prognosis in various cancers. To advance immunotherapies, testing on immunocompetent pre-clinical models of human tissue is crucial. We have developed an in vitro model of microvascular networks with tumor spheroids or patient tissues to assess monocyte trafficking into tumors and evaluate immunotherapies targeting the human tumor microenvironment. Our findings demonstrate that macrophages in vascularized breast and lung tumor models can enhance monocyte recruitment via CCL7 and CCL2, mediated by CSF-1R. Additionally, a multispecific antibody targeting CSF-1R, CCR2, and neutralizing TGF-ß (CSF1R/CCR2/TGF-ß Ab) repolarizes TAMs towards an anti-tumoral M1-like phenotype, reduces monocyte chemoattractant protein secretion, and blocks monocyte migration. This antibody also inhibits monocyte recruitment in patient-specific vascularized tumor models. In summary, this vascularized tumor model recapitulates the monocyte recruitment cascade, enabling functional testing of innovative therapeutic antibodies targeting TAMs in the tumor microenvironment.


Subject(s)
Monocytes , Receptor, Macrophage Colony-Stimulating Factor , Receptors, CCR2 , Tumor Microenvironment , Humans , Receptors, CCR2/metabolism , Receptors, CCR2/antagonists & inhibitors , Monocytes/metabolism , Monocytes/immunology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Female , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Mice , Cell Movement/drug effects , Neoplasms/immunology , Neoplasms/pathology
2.
Sci Immunol ; 9(99): eadp0344, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39241057

ABSTRACT

Langerhans cells (LCs) are distinct among phagocytes, functioning both as embryo-derived, tissue-resident macrophages in skin innervation and repair and as migrating professional antigen-presenting cells, a function classically assigned to dendritic cells (DCs). Here, we demonstrate that both intrinsic and extrinsic factors imprint this dual identity. Using ablation of embryo-derived LCs in the murine adult skin and tracking differentiation of incoming monocyte-derived replacements, we found intrinsic intraepidermal heterogeneity. We observed that ontogenically distinct monocytes give rise to LCs. Within the epidermis, Jagged-dependent activation of Notch signaling, likely within the hair follicle niche, provided an initial site of LC commitment before metabolic adaptation and survival of monocyte-derived LCs. In the human skin, embryo-derived LCs in newborns retained transcriptional evidence of their macrophage origin, but this was superseded by DC-like immune modules after postnatal expansion. Thus, adaptation to adult skin niches replicates conditioning of LC at birth, permitting repair of the embryo-derived LC network.


Subject(s)
Cell Differentiation , Langerhans Cells , Monocytes , Skin , Langerhans Cells/immunology , Langerhans Cells/cytology , Animals , Monocytes/immunology , Monocytes/cytology , Cell Differentiation/immunology , Humans , Skin/immunology , Skin/cytology , Mice , Mice, Inbred C57BL , Female
3.
Front Immunol ; 15: 1414716, 2024.
Article in English | MEDLINE | ID: mdl-39315092

ABSTRACT

Ovarian cancer (OC) is the most lethal gynecologic malignancy worldwide. The major clinical challenge includes the asymptomatic state of the disease, making diagnosis possible only at advanced stages. Another OC complication is the high relapse rate and poor prognosis following the standard first-line treatment with platinum-based chemotherapy. At present, numerous clinical trials are being conducted focusing on immunotherapy in OC; nevertheless, there are still no FDA-approved indications. Personalized decision regarding the immunotherapy, including immune checkpoint blockade and immune cell-based immunotherapies, can depend on the effective antigen presentation required for the cytotoxic immune response. The major aim of our study was to uncover tumor-specific transcriptional and epigenetic changes in peripheral blood monocytes in patients with high-grade serous ovarian cancer (HGSOC). Another key point was to elucidate how chemotherapy can reprogram monocytes and how that relates to changes in other immune subpopulations in the blood. To this end, we performed single-cell RNA sequencing of peripheral blood mononuclear cells (PBMCs) from patients with HGSOC who underwent neoadjuvant chemotherapeutic treatment (NACT) and in treatment-naïve patients. Monocyte cluster was significantly affected by tumor-derived factors as well as by chemotherapeutic treatment. Bioinformatical analysis revealed three distinct monocyte subpopulations within PBMCs based on feature gene expression - CD14.Mn.S100A8.9hi, CD14.Mn.MHC2hi and CD16.Mn subsets. The intriguing result was that NACT induced antigen presentation in monocytes by the transcriptional upregulation of MHC class II molecules, but not by epigenetic changes. Increased MHC class II gene expression was a feature observed across all three monocyte subpopulations after chemotherapy. Our data also demonstrated that chemotherapy inhibited interferon-dependent signaling pathways, but activated some TGFb-related genes. Our results can enable personalized decision regarding the necessity to systemically re-educate immune cells to prime ovarian cancer to respond to anti-cancer therapy or to improve personalized prescription of existing immunotherapy in either combination with chemotherapy or a monotherapy regimen.


Subject(s)
Antigen Presentation , Cystadenocarcinoma, Serous , Monocytes , Ovarian Neoplasms , Humans , Female , Monocytes/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/drug therapy , Antigen Presentation/drug effects , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/immunology , Middle Aged , Neoplasm Grading , Gene Expression Regulation, Neoplastic/drug effects , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoadjuvant Therapy/methods , Epigenesis, Genetic
4.
Med Microbiol Immunol ; 213(1): 19, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39297970

ABSTRACT

The Panton-Valentine leukocidin (PVL) of Staphylococcus aureus is associated with necrotizing infections. After binding to complement 5a receptor (C5aR/CD88) and CD45 it causes cytolysis in polymorphonuclear neutrophils (PMNs) as well as inflammasome activation in monocytes. The objective of this study was to test if (ant)agonists of C5aR and CD45 can attenuate the effect of PVL on PMNs and monocytes. We tested the effect of various concentrations of six C5aR (ant)agonists (avacopan, BM213, DF2593A, JPE-1375, PMX205 and W-54011) and one CD45 antagonist (NQ301) to attenuate the cytotoxic effect of PVL on human PMNs and monocytes in vitro. Shifts in the half-maximal effective concentration (EC50) of PVL to achieve a cytotoxic effect on PMNs and modulation of inflammatory cytokine response from monocytes were determined by flow cytometry and IL-1ß detection. Pre-treatment of PMNs with avacopan, PMX205 and W-54,011 resulted in 3.6- to 4.3-fold shifts in the EC50 for PVL and were able to suppress IL-1ß secretion by human monocytes in the presence of PVL. BM213, DF2593A and NQ301 were unable to change the susceptibility of PMNs towards PVL or reduce inflammasome activation in monocytes. Avacopan, PMX205 and W-54,011 showed protection against PVL-induced cytotoxicity and suppressed IL-1ß secretion by monocytes. Clinical studies are needed to prove whether these substances can be used therapeutically as repurposed drugs.


Subject(s)
Bacterial Toxins , Exotoxins , Leukocidins , Monocytes , Neutrophils , Receptor, Anaphylatoxin C5a , Staphylococcus aureus , Leukocidins/metabolism , Leukocidins/antagonists & inhibitors , Exotoxins/metabolism , Exotoxins/pharmacology , Exotoxins/antagonists & inhibitors , Humans , Bacterial Toxins/metabolism , Monocytes/drug effects , Monocytes/metabolism , Monocytes/immunology , Staphylococcus aureus/drug effects , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/metabolism , Leukocyte Common Antigens/metabolism , Anti-Bacterial Agents/pharmacology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Inflammasomes/metabolism , Interleukin-1beta/metabolism
5.
PLoS One ; 19(9): e0308644, 2024.
Article in English | MEDLINE | ID: mdl-39298419

ABSTRACT

Although it is widely known that various pharmaceuticals affect the methylome, the knowledge of the effects from anesthesia is limited, and nearly nonexistent regarding the effects of obstetric anesthesia on the newborn child. Using sequencing based-methylation data and a reference-based statistical deconvolution approach we performed methylome-wide association studies (MWAS) of neonatal whole blood, and for each cell-type specifically, to detect methylation variations that are associated with the pain relief administered to the mother during delivery. Significant findings were replicated in a different dataset and followed-up with gene ontology analysis to pinpoint biological functions of potential relevance to these neonatal methylation alterations. The MWAS analyses detected methylome-wide significant (q<0.1) alterations in the newborn for laughing gas in granulocytes (two CpGs, p<5.50x10-9, q = 0.067), and for pudendal block in monocytes (five CpGs across three loci, p<1.51 x10-8, q = 0.073). Suggestively significant findings (p<1.00x10-6) were detected for both treatments for bulk and all cell-types, and replication analyses showed consistent significant enrichment (odds ratios ranging 3.47-39.02; p<4.00×10-4) for each treatment, suggesting our results are robust. In contrast, we did not observe any overlap across treatments, suggesting that the treatments are associated with different alterations of the neonatal blood methylome. Gene ontology analyses of the replicating suggestively significant results indicated functions related to, for example, cell differentiation, intracellular membrane-bound organelles and calcium transport. In conclusion, for the first time, we investigated and detected effect of obstetric pain-relief on the blood methylome in the newborn child. The observed differences suggest that anesthetic treatment, such as laughing gas or pudendal block, may alter the neonatal methylome in a cell-type specific manner. Some of the observed alterations are part of gene ontology terms that previously have been suggested in relation to anesthetic treatment, supporting its potential role also in obstetric anesthesia.


Subject(s)
DNA Methylation , Humans , Infant, Newborn , Female , Pregnancy , Genome-Wide Association Study , CpG Islands , Monocytes/metabolism , Pain Management/methods , Epigenome
6.
Bull Exp Biol Med ; 177(4): 423-426, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39264558

ABSTRACT

The level of ROS (fluorescent probe 2',7'-dichlorodihydrofluorescein diacetate) and lipid content (fluorescent lipophilic dye Nile Red) in the peripheral blood monocyte fraction from patients with type 1 diabetes mellitus and healthy volunteers were assessed by flow cytofluorimetry. The number of CD36+ monocytes was assessed using specific antibodies. In patients with type 1 diabetes mellitus, the levels of ROS and intracellular lipids in monocytes and the number of cells expressing CD36 fatty acid translocase were elevated. These results indicate metabolic changes in the peripheral blood cells of patients with carbohydrate metabolism disorders and can be considered as possible prognostic markers for the development of type 1 diabetes mellitus complications.


Subject(s)
Diabetes Mellitus, Type 1 , Monocytes , Reactive Oxygen Species , Humans , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/metabolism , Monocytes/metabolism , Male , Adult , Female , Reactive Oxygen Species/metabolism , CD36 Antigens/metabolism , CD36 Antigens/blood , Case-Control Studies , Flow Cytometry , Young Adult , Lipid Metabolism
7.
Tuberk Toraks ; 72(3): 197-207, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39275932

ABSTRACT

Introduction: Platelets (PLT) and host systemic inflammatory response (SIR) are known to be effective in the aggregation of cancer cells and the formation of metastasis. There are studies pointing out to the prognostic efficacy of lymphocyte-monocyte ratio (LMR) showing SIR activation and mean platelet volume (MPV) values indicating platelet activation in various cancer types. We predict that easy-to-access hemogram parameters such as MPV, MPV/PLT, and LMR can be guiding in the clinical follow-up period of patients with epidermal growth factor receptor (EGFR) positive mutation and who received EGFR, tyrosine kinase inhibitor (TKI) in the first-line treatment in predicting the progression of the disease, predicting the survival time of the patients, and evaluating the response to treatment. Materials and Methods: The study is retrospective and included patients with stage III and stage IV pulmonary adenocarcinoma with positive EGFR mutations and for whom TKI was used in the first-line treatment between January 2011 and January 2021. MPV, MPV/PLT, and LMR values of the patients were calculated before treatment. Age, sex, comorbidity, smoking history, TNM stage, metastasis localizations, EGFR mutation types, TKI treatments used in first-line treatment, and MPV, MPV/PLT, and LMR values at the 1st month of treatment were recorded. With Kaplan-Meier, six-month, one-year, three-year, and five-year survival rates, average life expectancy, and 95% confidence intervals for these periods were calculated. Variables that may affect progression and overall survival (OS) were determined by performing univariate and multivariate Cox regression analysis. Result: One hundred and two patients were included in the study. The mean age of the patients was 64.30 ± 12.6 years. Eighty-four patients were in stage IV at the time of diagnosis. The expected mean progression-free survival (PFS) period of the cases was found to be 13.3 months. The mean life expectancy of the cases was found to be 35.1 months. Web-based Cutoff Finder algorithm written in the R program (http://molpath.charite.de/cutoff) was used to determine the ideal cut points for MPV, MPV/PLT, and LMR. The cut-off values were found to be 7.55 fL for MPV, 0.251 for MPV/PLT, and 2.615 for LMR, respectively. In univariate Cox regression analysis, LMR level lower than 2.615 increased the rate of progression 1.747 times (95% confidence interval: 1.129-2.705) and the death rate 2.056 times (95% confidence interval: 1.217-3.475) (p= 0.012, p= 0.007). The mean PFS LMR cut-off value was 10.3 months, and 15.3 months, and mean OS durations were 25.1 months and 40.8 months for the groups with low and high cut-off values respectively (p= 0.011, p= 0.006 log-rank test). According to the results of multivariate Cox regression analysis, MPV/PLT < 0.251, smoking, presence of pleural and adrenal metastases, and gefitinib treatment were independent factors in determining PFS. The independent factors determining OS in multivariate Cox regression analysis were being male, platelet increase, MPV > 7.55, gefitinib treatment, and smoking. Conclusions: MPV, MPV/PLT, and LMR are potential biomarkers that can be used for the clinical follow-up of lung ADC patients receiving EGFR-TKI treatment.


Subject(s)
Adenocarcinoma of Lung , ErbB Receptors , Lung Neoplasms , Mean Platelet Volume , Protein Kinase Inhibitors , Humans , Male , Female , Middle Aged , ErbB Receptors/genetics , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/blood , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Retrospective Studies , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/blood , Adenocarcinoma of Lung/pathology , Aged , Protein Kinase Inhibitors/therapeutic use , Prognosis , Mutation , Monocytes , Blood Platelets , Lymphocytes
8.
Biomater Adv ; 165: 214024, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39232353

ABSTRACT

Graphene oxide (GO), a carbon-based nanomaterial, presents significant potential across biomedical fields such as bioimaging, drug delivery, biosensors, and phototherapy. This study examines the effects of integrating GO into poly(lactic-co-glycolic acid) (PLGA) scaffolds on human immune cell function. Our results demonstrate that high concentrations of GO reduce the viability of peripheral blood mononuclear cells (PBMCs) following stimulation with anti-CD3 antibody. This reduction extends to T lymphocyte activation, evident from the diminished proliferative response to T cell receptor engagement and impaired differentiation into T helper subsets and regulatory T cells. Interestingly, although GO induces a minimal response in resting monocytes, but it significantly affects both the viability and the differentiation potential of monocytes induced to mature toward M1 pro-inflammatory and M2-like immunoregulatory macrophages. This study seeks to address a critical gap by investigating the in vitro immunomodulatory effects of PLGA scaffolds incorporating various concentrations of GO on primary immune cells, specifically PBMCs isolated from healthy donors. Our findings emphasize the need to optimize the GO to PLGA ratios and scaffold design to advance PLGA-GO-based biomedical applications. STATEMENT OF SIGNIFICANCE: Graphene oxide (GO) holds immense promise for biomedical applications due to its unique properties. However, concerns regarding its potential to trigger adverse immune responses remain. This study addresses this critical gap by investigating the in vitro immunomodulatory effects of PLGA scaffolds incorporating increasing GO concentrations on human peripheral blood mononuclear cells (PBMCs). By elucidating the impact on cell viability, T cell proliferation and differentiation, and the maturation/polarization of antigen-presenting cells, this work offers valuable insights for designing safe and immunologically compatible GO-based biomaterials for future clinical translation.


Subject(s)
Graphite , Leukocytes, Mononuclear , Polylactic Acid-Polyglycolic Acid Copolymer , Tissue Scaffolds , Graphite/chemistry , Graphite/pharmacology , Humans , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Tissue Scaffolds/chemistry , Cell Differentiation/drug effects , Cell Survival/drug effects , Cell Proliferation/drug effects , Lymphocyte Activation/drug effects , Biocompatible Materials/pharmacology , Biocompatible Materials/chemistry , Monocytes/drug effects , Monocytes/immunology , Macrophages/drug effects , Macrophages/immunology
9.
Int J Mol Sci ; 25(17)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39273110

ABSTRACT

The role of immune system components in the development of myocardial remodeling in chronic kidney disease (CKD) and kidney transplantation remains an open question. Our aim was to investigate the associations between immune cell subpopulations in the circulation of CKD patients and kidney transplant recipients (KTRs) with subclinical indices of myocardial performance. We enrolled 44 CKD patients and 38 KTRs without established cardiovascular disease. A selected panel of immune cells was measured by flow cytometry. Classical and novel strain-related indices of ventricular function were measured by speckle-tracking echocardiography at baseline and following dipyridamole infusion. In CKD patients, the left ventricular (LV) relative wall thickness correlated with the CD14++CD16- monocytes (ß = 0.447, p = 0.004), while the CD14++CD16+ monocytes were independent correlates of the global radial strain (ß = 0.351, p = 0.04). In KTRs, dipyridamole induced changes in global longitudinal strain correlated with CD14++CD16+ monocytes (ß = 0.423, p = 0.009) and CD4+ T-cells (ß = 0.403, p = 0.01). LV twist and untwist were independently correlated with the CD8+ T-cells (ß = 0.405, p = 0.02 and ß = -0.367, p = 0.03, respectively) in CKD patients, whereas the CD14++CD16+ monocytes were independent correlates of LV twist and untwist in KTRs (ß = 0.405, p = 0.02 and ß = -0.367, p = 0.03, respectively). Immune cell subsets independently correlate with left ventricular strain and torsion-related indices in CKD patients and KTRs without established CVD.


Subject(s)
Kidney Transplantation , Monocytes , Renal Insufficiency, Chronic , Humans , Kidney Transplantation/adverse effects , Male , Female , Renal Insufficiency, Chronic/immunology , Middle Aged , Monocytes/metabolism , Monocytes/immunology , Echocardiography , Adult , Lipopolysaccharide Receptors/metabolism , Cardiovascular Diseases/etiology , Aged , Transplant Recipients , Immune System , Receptors, IgG/metabolism
10.
Int J Mol Sci ; 25(17)2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39273424

ABSTRACT

Communication between natural killer cells (NK cells) and monocytes/macrophages may play an important role in immunomodulation and regulation of inflammatory processes. The aim of this research was to investigate the impact of NK cell-derived large extracellular vesicles on monocyte function because this field is understudied. We studied how NK-cell derived large extracellular vesicles impact on THP-1 cells characteristics after coculturing: phenotype, functions were observed with flow cytometry. In this study, we demonstrated the ability of large extracellular vesicles produced by NK cells to integrate into the membranes of THP-1 cells and influence the viability, phenotype, and functional characteristics of the cells. The results obtained demonstrate the ability of large extracellular vesicles to act as an additional component in the immunomodulatory activity of NK cells in relation to monocytes.


Subject(s)
Extracellular Vesicles , Killer Cells, Natural , Monocytes , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Monocytes/immunology , Monocytes/metabolism , Monocytes/cytology , THP-1 Cells , Coculture Techniques , Cell Communication/immunology , Cell Survival , Macrophages/immunology , Macrophages/metabolism
11.
Int J Mol Sci ; 25(17)2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39273504

ABSTRACT

In recent years, Raman spectroscopy has garnered growing interest in the field of biomedical research. It offers a non-invasive and label-free approach to defining the molecular fingerprint of immune cells. We utilized Raman spectroscopy on optically trapped immune cells to investigate their molecular compositions. While numerous immune cell types have been studied in the past, the characterization of living human CD3/CD28-stimulated T cell subsets remains incomplete. In this study, we demonstrate the capability of Raman spectroscopy to readily distinguish between naïve and stimulated CD4 and CD8 cells. Additionally, we compared these cells with monocytes and discovered remarkable similarities between stimulated T cells and monocytes. This paper contributes to expanding our knowledge of Raman spectroscopy of immune cells and serves as a launching point for future clinical applications.


Subject(s)
Monocytes , Spectrum Analysis, Raman , T-Lymphocyte Subsets , Humans , Spectrum Analysis, Raman/methods , Monocytes/cytology , Monocytes/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Optical Tweezers , CD8-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Lymphocyte Activation , CD28 Antigens/metabolism , CD28 Antigens/immunology
12.
Nat Commun ; 15(1): 7863, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251592

ABSTRACT

Regulatory T cells (Tregs) are crucial immune cells for tissue repair and regeneration. However, their potential as a cell-based regenerative therapy is not yet fully understood. Here, we show that local delivery of exogenous Tregs into injured mouse bone, muscle, and skin greatly enhances tissue healing. Mechanistically, exogenous Tregs rapidly adopt an injury-specific phenotype in response to the damaged tissue microenvironment, upregulating genes involved in immunomodulation and tissue healing. We demonstrate that exogenous Tregs exert their regenerative effect by directly and indirectly modulating monocytes/macrophages (Mo/MΦ) in injured tissues, promoting their switch to an anti-inflammatory and pro-healing state via factors such as interleukin (IL)-10. Validating the key role of IL-10 in exogenous Treg-mediated repair and regeneration, the pro-healing capacity of these cells is lost when Il10 is knocked out. Additionally, exogenous Tregs reduce neutrophil and cytotoxic T cell accumulation and IFN-γ production in damaged tissues, further dampening the pro-inflammatory Mo/MΦ phenotype. Highlighting the potential of this approach, we demonstrate that allogeneic and human Tregs also promote tissue healing. Together, this study establishes exogenous Tregs as a possible universal cell-based therapy for regenerative medicine and provides key mechanistic insights that could be harnessed to develop immune cell-based therapies to enhance tissue healing.


Subject(s)
Interleukin-10 , Macrophages , Mice, Inbred C57BL , T-Lymphocytes, Regulatory , Wound Healing , Animals , T-Lymphocytes, Regulatory/immunology , Wound Healing/immunology , Interleukin-10/metabolism , Interleukin-10/genetics , Humans , Mice , Macrophages/immunology , Male , Monocytes/immunology , Skin/immunology , Interferon-gamma/metabolism , Interferon-gamma/immunology , Female
13.
Cell Biochem Funct ; 42(7): e4115, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39264203

ABSTRACT

In this study, the protective effects of Panax notoginseng saponins (PNS) against gamma radiation-induced DNA damage and associated physiological alterations in Swiss albino mice were investigated. Exposure to gamma radiation led to a dose-dependent increase in cytokinesis-blocked micronuclei (CBMN) double-strand DNA breaks (DSBs), dicentric aberrations (DC), formation in peripheral blood mononuclear cells. However, pretreatment with PNS at concentrations of 1, 5, and 10 µg/mL significantly attenuated the frequencies of DC and CBMN in a concentration-dependent manner. PNS administration before radiation exposure also reduced radiation-induced DSBs in BL, indicating protection against reactive oxygen species generation and DNA damage. Notably, pretreatment with PNS at 10 µg/mL prevented the overexpression of γ-H2AX, proteins associated with DNA damage response, in irradiated mice. In addition, in vivo studies showed intraperitoneal administration of PNS (25 mg/kg body weight) for 1 h before radiation exposure mitigated lipid peroxidation levels and restored antioxidant status, countering oxidative damage induced by gamma radiation. Furthermore, PNS pretreatment reversed the decrease in hemoglobin (Hb) content, white blood cell count, and red blood cell count in irradiated mice, indicating preservation of hematological parameters. Overall, PNS demonstrated an anticlastogenic effect by modulating radiation-induced DSBs and preventing oxidative damage, thus highlighting its potential as a protective agent against radiation-induced DNA damage and associated physiological alterations. Clinically, PNS will be beneficial for cancer patients undergoing radiotherapy, but their pharmacological properties and toxicity profiles need to be studied.


Subject(s)
Gamma Rays , Panax notoginseng , Saponins , Animals , Gamma Rays/adverse effects , Saponins/pharmacology , Mice , Panax notoginseng/chemistry , Humans , Male , DNA Damage/drug effects , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/radiation effects , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Radiation-Protective Agents/pharmacology , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , Monocytes/drug effects , Monocytes/metabolism , Monocytes/radiation effects , Reactive Oxygen Species/metabolism , Antioxidants/pharmacology
14.
JCI Insight ; 9(17)2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39253970

ABSTRACT

HIV-associated neurocognitive impairment (HIV-NCI) affects 15%-50% of people with HIV (PWH), despite viral suppression with antiretroviral therapy (ART). HIV neuropathogenesis is mediated, in part, by transmigration of infected CD14+CD16+ monocytes across the blood-brain barrier (BBB) into the central nervous system (CNS). In the CNS, CD14+CD16+ monocytes contribute to infection and activation of parenchymal cells, resulting in production of neurotoxic viral and host factors that cause neuronal damage. Mechanisms by which CD14+CD16+ monocytes contribute to HIV-NCI have not been characterized in a study population of PWH on ART without contribution from confounders that affect cognition (e.g., substance use, hepatitis C virus coinfection). We assessed cognitive function, PBMC transmigration across the BBB, and neuronal health markers in a well-defined cohort of 56 PWH on ART using stringent criteria to eliminate confounding factors. We demonstrated that PWH on ART with HIV-NCI have significantly increased transmigration of their CD14+CD16+ monocytes across the BBB compared with those with normal cognition. We showed that hypertension and diabetes may be effect modifiers on the association between CD14+CD16+ monocyte transmigration and cognition. This study underscored the persistent role of CD14+CD16+ monocytes in HIV-NCI, even in PWH with viral suppression, suggesting them as potential targets for therapeutic interventions.


Subject(s)
Blood-Brain Barrier , HIV Infections , Lipopolysaccharide Receptors , Monocytes , Receptors, IgG , Humans , Blood-Brain Barrier/metabolism , Receptors, IgG/metabolism , Monocytes/metabolism , Monocytes/immunology , Lipopolysaccharide Receptors/metabolism , Male , Female , Middle Aged , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , HIV Infections/metabolism , Adult , GPI-Linked Proteins/metabolism , AIDS Dementia Complex/immunology , AIDS Dementia Complex/metabolism
15.
J Cell Mol Med ; 28(18): e18507, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39288445

ABSTRACT

The potential of extracellular vesicles (EVs) isolated from mesenchymal stromal cells in guiding macrophages toward anti-inflammatory immunophenotypes, has been reported in several studies. In our study, we provided experimental evidence of a distinctive effect played by Wharton Jelly mesenchymal stromal cell-derived EVs (WJ-EVs) on human macrophages. We particularly analyzed their anti-inflammatory effects on macrophages by evaluating their interactions with stellate cells, and their protective role in liver fibrosis. A three-step gradient method was used to isolate monocytes from umbilical cord blood (UCB). Two subpopulations of WJ-EVs were isolated by high-speed (20,000 g) and differential ultracentrifugation (110,000 g). Further to their characterization, they were designated as EV20K and EV110K and incubated at different concentrations with UCB-derived monocytes for 7 days. Their anti-fibrotic effect was assessed by studying the differentiation and functional levels of generated macrophages and their potential to modulate the survival and activity of LX2 stellate cells. The EV20K triggers the polarization of UCB-derived monocytes towards a peculiar M2-like functional phenotype more effectively than the M-CSF positive control. The EV20K treated macrophages were characterized by a higher expression of scavenger receptors, increased phagocytic capacity and production level of interleukin-10 and transforming growth factor-ß. Conditioned medium from those polarized macrophages attenuated the proliferation, contractility and activation of LX2 stellate cells. Our data show that EV20K derived from WJ-MSCs induces activated macrophages to suppress immune responses and potentially play a protective role in the pathogenesis of liver fibrosis by directly inhibiting HSC's activation.


Subject(s)
Cell Differentiation , Extracellular Vesicles , Liver Cirrhosis , Macrophages , Mesenchymal Stem Cells , Phenotype , Wharton Jelly , Mesenchymal Stem Cells/metabolism , Humans , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Extracellular Vesicles/metabolism , Macrophages/metabolism , Wharton Jelly/cytology , Macrophage Activation , Hepatic Stellate Cells/metabolism , Monocytes/metabolism , Phagocytosis , Fetal Blood/cytology , Fetal Blood/metabolism
16.
Int J Mol Sci ; 25(17)2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39273692

ABSTRACT

Understanding the pig immune function is crucial for disease-resistant breeding and potentially for human health research due to shared immune system features. Immune cell ratios, like monocyte/lymphocyte ratio (MLR) and neutrophil/lymphocyte ratio (NLR), offer a more comprehensive view of immune status compared to individual cell counts. However, research on pig immune cell ratios remains limited. This study investigated MLR and NLR in a Duroc × Erhualian F2 resource population. Heritability analysis revealed high values (0.649 and 0.688 for MLR and NLR, respectively), suggesting a strong genetic component. Furthermore, we employed an ensemble-like GWAS (E-GWAS) strategy and functional annotation analysis to identify 11 MLR-associated and 6 NLR-associated candidate genes. These genes were significantly enriched in immune-related biological processes. These findings provide novel genetic markers and candidate genes associated with porcine immunity, thereby providing valuable insights for addressing biosecurity and animal welfare concerns in the pig industry.


Subject(s)
Lymphocytes , Monocytes , Neutrophils , Polymorphism, Single Nucleotide , Animals , Monocytes/metabolism , Lymphocytes/metabolism , Lymphocytes/immunology , Neutrophils/metabolism , Neutrophils/immunology , Swine , Genome-Wide Association Study , Male , Female , Leukocyte Count
17.
Nat Commun ; 15(1): 7662, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266531

ABSTRACT

Most patients with advanced cancer develop cachexia, a multifactorial syndrome characterized by progressive skeletal muscle wasting. Despite its catastrophic impact on survival, the critical mediators responsible for cancer cachexia development remain poorly defined. Here, we show that a distinct subset of neutrophil-like monocytes, which we term cachexia-inducible monocytes (CiMs), emerges in the advanced cancer milieu and promotes skeletal muscle loss. Unbiased transcriptome analysis reveals that interleukin 36 gamma (IL36G)-producing CD38+ CiMs are induced in chronic monocytic blood cancer characterized by prominent cachexia. Notably, the emergence of CiMs and the activation of CiM-related gene signatures in monocytes are confirmed in various advanced solid cancers. Stimuli of toll-like receptor 4 signaling are responsible for the induction of CiMs. Genetic inhibition of IL36G-mediated signaling attenuates skeletal muscle loss and rescues cachexia phenotypes in advanced cancer models. These findings indicate that the IL36G-producing subset of neutrophil-like monocytes could be a potential therapeutic target in cancer cachexia.


Subject(s)
Cachexia , Monocytes , Muscle, Skeletal , Neoplasms , Neutrophils , Cachexia/metabolism , Cachexia/etiology , Monocytes/metabolism , Monocytes/immunology , Humans , Neoplasms/complications , Neoplasms/metabolism , Neoplasms/immunology , Neutrophils/metabolism , Animals , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mice , Male , Signal Transduction , Cell Line, Tumor , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Mice, Inbred C57BL , Interleukins/metabolism , Interleukins/genetics , Female , Gene Expression Profiling
18.
Nat Commun ; 15(1): 7998, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266542

ABSTRACT

Impaired clearance of amyloid ß (Aß) in late-onset Alzheimer's disease (AD) affects disease progression. The role of peripheral monocytes in Aß clearance from the central nervous system (CNS) is unclear. We use a flow cytometry assay to identify Aß-binding monocytes in blood, validated by confocal microscopy, Western blotting, and mass spectrometry. Flow cytometry immunophenotyping and correlation with AD biomarkers are studied in 150 participants from the AIBL study. We also examine monocytes in human cerebrospinal fluid (CSF) and their migration in an APP/PS1 mouse model. The assay reveals macrophage-like Aß-binding monocytes with high phagocytic potential in both the periphery and CNS. We find lower surface Aß levels in mild cognitive impairment (MCI) and AD-dementia patients compared to cognitively unimpaired individuals. Monocyte infiltration from blood to CSF and migration from CNS to peripheral lymph nodes and blood are observed. Here we show that Aß-binding monocytes may play a role in CNS Aß clearance, suggesting their potential as a biomarker for AD diagnosis and monitoring.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Cognitive Dysfunction , Disease Progression , Mice, Transgenic , Monocytes , Alzheimer Disease/metabolism , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Alzheimer Disease/blood , Humans , Monocytes/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Female , Aged , Male , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/cerebrospinal fluid , Mice , Aged, 80 and over , Biomarkers/cerebrospinal fluid , Biomarkers/blood , Biomarkers/metabolism , Flow Cytometry , Disease Models, Animal , Phagocytosis , Middle Aged
19.
Nature ; 633(8028): 155-164, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39232147

ABSTRACT

Infectious, inflammatory and autoimmune conditions present differently in males and females. SARS-CoV-2 infection in naive males is associated with increased risk of death, whereas females are at increased risk of long COVID1, similar to observations in other infections2. Females respond more strongly to vaccines, and adverse reactions are more frequent3, like most autoimmune diseases4. Immunological sex differences stem from genetic, hormonal and behavioural factors5 but their relative importance is only partially understood6-8. In individuals assigned female sex at birth and undergoing gender-affirming testosterone therapy (trans men), hormone concentrations change markedly but the immunological consequences are poorly understood. Here we performed longitudinal systems-level analyses in 23 trans men and found that testosterone modulates a cross-regulated axis between type-I interferon and tumour necrosis factor. This is mediated by functional attenuation of type-I interferon responses in both plasmacytoid dendritic cells and monocytes. Conversely, testosterone potentiates monocyte responses leading to increased tumour necrosis factor, interleukin-6 and interleukin-15 production and downstream activation of nuclear factor kappa B-regulated genes and potentiation of interferon-γ responses, primarily in natural killer cells. These findings in trans men are corroborated by sex-divergent responses in public datasets and illustrate the dynamic regulation of human immunity by sex hormones, with implications for the health of individuals undergoing hormone therapy and our understanding of sex-divergent immune responses in cisgender individuals.


Subject(s)
Testosterone , Transgender Persons , Adult , Female , Humans , Male , Datasets as Topic , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/drug effects , Immune System/drug effects , Immune System/metabolism , Interferon Type I/immunology , Interferon Type I/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-15/immunology , Interleukin-15/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/drug effects , Monocytes/immunology , Monocytes/drug effects , Monocytes/metabolism , NF-kappa B/metabolism , Sex Characteristics , Testosterone/adverse effects , Testosterone/immunology , Testosterone/pharmacology , Testosterone/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
20.
Lung ; 202(5): 683-693, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39284999

ABSTRACT

INTRODUCTION: In sarcoidosis granulomas, monocyte-derived macrophages are activated by pro-inflammatory cytokines including TNF and IL-6. Current drug treatment for sarcoidosis aims to suppress inflammation but disabling side effects can ensue. The macrolide azithromycin may be anti-inflammatory. We aimed to determine whether treatment with azithromycin affects blood inflammatory gene expression and monocyte functions in sarcoidosis. METHODS: Blood samples were collected from patients with chronic pulmonary sarcoidosis enrolled in a single arm, open label clinical trial who received oral azithromycin 250 mg once daily for 3 months. Whole blood inflammatory gene expression with or without LPS stimulation was measured using a 770-mRNA panel. Phenotypic analysis and cytokine production were conducted by flow cytometry and ELISA after 24h stimulation with growth factors and TLR ligands. mTOR activity was assessed by measuring phosphorylated S6RP. RESULTS: Differential gene expression analysis indicated a state of heightened myeloid cell activation in sarcoidosis. Compared with controls, sarcoidosis patients showed increased LPS responses for several cytokines and chemokines. Treatment with azithromycin had minimal effect on blood gene expression overall, but supervised clustering analysis identified several chemokine genes that were upregulated. At the protein level, azithromycin treatment increased LPS-stimulated TNF and unstimulated IL-8 production. No other cytokines showed significant changes following azithromycin. Blood neutrophil counts fell during azithromycin treatment whereas mononuclear cells remained stable. Azithromycin had no detectable effects on mTOR activity or activation markers. CONCLUSION: Blood myeloid cells are activated in sarcoidosis, but azithromycin therapy did not suppress inflammatory gene expression or cytokine production in blood. TRIAL REGISTRATION: EudraCT 2019-000580-24 (17 May 2019).


Subject(s)
Azithromycin , Cytokines , Sarcoidosis, Pulmonary , Humans , Azithromycin/therapeutic use , Azithromycin/pharmacology , Middle Aged , Female , Male , Sarcoidosis, Pulmonary/drug therapy , Sarcoidosis, Pulmonary/blood , Cytokines/blood , Cytokines/genetics , Adult , Interleukin-8/blood , Interleukin-8/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/blood , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Monocytes/metabolism , Gene Expression/drug effects , Aged , Inflammation Mediators/blood , Inflammation Mediators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL